The role of mammalian poly(A)-binding proteins in co-ordinating mRNA turnover

Page created by Angel Sutton
 
CONTINUE READING
The role of mammalian poly(A)-binding proteins in co-ordinating mRNA turnover
856   Biochemical Society Transactions (2012) Volume 40, part 4

      The role of mammalian poly(A)-binding proteins
      in co-ordinating mRNA turnover
      Matthew Brook and Nicola K. Gray1
      MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, U.K.

                       Abstract
                       The function of cytoplasmic PABPs [poly(A)-binding proteins] in promoting mRNA translation has been
                       intensively studied. However, PABPs also have less clearly defined functions in mRNA turnover including
                       roles in default deadenylation, a major rate-limiting step in mRNA decay, as well as roles in the regulation
                       of mRNA turnover by cis-acting control elements and in the detection of aberrant mRNA transcripts. In
                       the present paper, we review our current understanding of the complex roles of PABP1 in mRNA turnover,
                       focusing on recent progress in mammals and highlighting some of the major questions that remain to be
                       addressed.

      Introduction                                                                                            absence of cis-acting regulatory elements, so-called default
      The extent to which an mRNA is utilized is determined by its                                            deadenylation occurs. Initial poly(A) removal occurs at a
      lifespan and rate of translation. These are intimately linked                                           constant low rate due to the distributive nature of PAN2–
      and can be regulated by cis-acting elements in response to                                              PAN3 which has a high on/off rate of RNA binding, therefore
      the cellular context. In eukaryotes, the primary determinants                                           only removing exposed poly(A). In contrast, CCR4–CAF1
      of both mRNA translation and turnover are the 5 m7 GTP                                                 acts processively, presumably evicting PABP1, and rapidly
      (7-methylguanosine triphosphate) ‘cap’ and 3 poly(A) tail                                              removes the remaining ∼110 nucleotides to leave an oligo(A)
      which act by binding eIF (eukaryotic initiation factor) 4F                                              tail. This oligo(A) tail is bound by the LSm (like-Sm) 1-7–Pat1
      (comprising eIF4E, eIF4G and eIF4A) and PABPs [poly(A)-                                                 complex which promotes removal of the 5 cap (decapping)
      binding proteins] respectively. The association of these                                                by the DCP (decapping protein) 1–DCP2 complex, allowing
      5 - and 3 -bound factors, primarily through eIF4G–PABP                                                access of the 5 →3 exonuclease XRN1 (exoribonuclease 1)
      interactions, forms a ‘closed-loop’ mRNA conformation                                                   which degrades the body of the mRNA in concert with 3 →5
      (Figure 1). This enhances ribosomal recruitment [1] and                                                 exonucleolytic activities (i.e. exosome and/or the LSm1-7–
      protects both ends of the mRNA, most likely by increasing                                               Pat1 complex) [3–5].
      binding of these factors to block access of the mRNA                                                       Cytoplasmic PABPs are conserved across eukaryotes and
      degradation machinery.                                                                                  are best characterized as central regulators of mRNA trans-
          mRNA turnover is multi-step process in which deadenyla-                                             lation (reviewed in [6–8]). Although mammals encode five
      tion [shortening of the poly(A) tail] is usually the initial                                            PABPs, almost all studies have focused on the prototypical
      and primary rate-limiting step. In mammals, deadenylation                                               family member, PABP1 (also known as PABPC1). PABP1
      is a biphasic process where poly(A) tails are shortened to                                              contains four non-identical RRMs (RNA-recognition motifs)
      ∼110 nucleotides (from approximately 250 nucleotides) by                                                and a C-terminal region. RRMs 1 and 2 bind poly(A) with
      the PAN [poly(A) nuclease] 2–PAN3 deadenylase complex                                                   high specificity, whereas RRMs 3 and 4 bind poly(A)
      and subsequently to oligo(A) (approximately A10–18 ) by                                                 with reduced affinity and can bind adenine/uridine-rich
      the CCR4 (carbon catabolite repressor protein 4)–CAF1                                                   RNA, although their RNA-binding specificity remains less
      (CCR4-associated factor 1) complex (reviewed in [2]). In the                                            clear [6]. Interestingly, the RRMs also mediate important
                                                                                                              protein interactions, including those with eIF4G [8].
                                                                                                              The C-terminal region is composed of the PABC (PABP
      Key words: cytoplasmic poly(A)-binding protein (PABP), deadenylation, microRNA (miRNA),
      mRNA decay, nonsense-mediated decay, translation factor.
                                                                                                              C-terminal domain)/MLLE domain which interacts with
      Abbreviations used: ARE, adenine/uridine-rich element; AUBP, ARE-binding protein; AUF1,                 numerous PAM2 (PABP-interacting motif 2)-containing
      adenine/uridine-rich RNA-binding factor 1; CCR4, carbon catabolite repressor protein 4; CAF1,           proteins {e.g. eRF (eukaryotic release factor) 3 [8],
      CCR4-associated factor 1; CRE, cytosine-rich element; DCP, decapping protein; eIF, eukaryotic
      initiation factor; EJC, exon-junction complex; ErEN, erythroid-cell-specific endonuclease; eRF,
                                                                                                              deadenylases [2,9,10] and TNRC6 (trinucleotide repeat-
      eukaryotic release factor; LSm, like-Sm; mCRD, major coding-region determinant; miRNA,                  containing 6) [11]}, and a proline-rich linker that is involved
      microRNA; NF-L, neurofilament light; NMD, nonsense-mediated decay; NOT, negative on TATA;                in PABP1–PABP1 interactions which enhance co-operative
      NSAP1, NS1-associated protein 1; PABP, poly(A)-binding protein; ePAPB, embryonic PABP; PABC,
      PABP C-terminal domain; PAIP1, PABP-interacting protein 1; PAM2, PABP-interacting motif 2;
                                                                                                              poly(A) binding [6,12,13]. The known protein partners of
      PAN, poly(A) nuclease; PARN, poly(A)-specific ribonuclease; PTC, premature termination codon;            PABP1 provide some insight into its complex functions in
      PTM, post-translational modification; RISC, RNA-induced silencing complex; RRM, RNA-recognition          both mRNA translation and stability, and in the present paper
      motif; TNRC6, trinucleotide repeat-containing 6; TOB, transducer of ErbB2; TTP, tristetraprolin; UNR,
      upstream of N-ras; UPF, up-frameshift; UTR, untranslated region; XRN1, exoribonuclease 1.
                                                                                                              we review the current state of knowledge regarding its role in
      1
        To whom correspondence should be addressed (email Nicola.Gray@ed.ac.uk).                              regulating mRNA turnover in mammals.

      
      C The   Authors Journal compilation 
                                          C 2012 Biochemical Society                                                        Biochem. Soc. Trans. (2012) 40, 856–864; doi:10.1042/BST20120100
The role of mammalian poly(A)-binding proteins in co-ordinating mRNA turnover
RNA UK 2012     857

Figure 1 Role of PABP1 in the closed-loop mRNA conformation                  the CCR4–NOT–CAF1 complex via TOB (transducer of
mRNAs are bound at the cap by eIF4E, as part of the eIF4F complex (eIF4E,    ErbB2) 1 (or TOB2) [2]. Somewhat counterintuitively,
eIF4G, eIF4A) and at the poly(A) tail by PABP1, protecting the mRNA          given its poly(A)-protective effect, PABP1 also appears
ends from decapping and deadenylation. Both eIF4E and PABP1 bind the         to enhance PAN2–PAN3 complex activity and, via TOB-
scaffolding protein eIF4G, which effectively bridges the 5 and 3 ends of   mediated recruitment, CCR4–CAF1 deadenylase activity
the mRNA to form a ‘closed-loop’. This conformation increases the affinity    [2,17] (Figure 2, panel 1). Such observations, although
of eIF4E for the cap and is proposed to increase the affinity of PABP1        confusing, support complex roles for PABP1 as both as
for the poly(A) tail, promoting small ribosomal subunit recruitment [1]      a facilitator of deadenylation and as a negative regulator
and further protecting mRNAs from decay (not depicted). For simplicity,      of deadenylation, with its interactions with deadenylases
other initiation factors that interact with PABP1, including those that      raising the possibility that PABP1 may inhibit their
contribute to the closed-loop, are not shown.                                function directly as well as sterically. Since it was proposed
                                                                             recently that only non-co-operatively bound PABP1 may
                                                                             be available for PABC-mediated interactions [12], it is
                                                                             tempting to speculate that co-operatively bound PABP1
                                                                             acts as a steric block with non-co-operatively bound
                                                                             PABP1 recruiting/stimulating deadenylases.

                                                                             Might PABP1 be removed from poly(A) tails
                                                                             during translation termination?
                                                                             Work in yeast first highlighted potential links between
                                                                             deadenylation and translational termination, by showing that
                                                                             the translation termination factor eRF3 interacts with PAB-1.
                                                                             This led to a model in which eRF3–PAB-1 interaction during
                                                                             termination facilitates the removal of a PAB-1 molecule from
                                                                             the poly(A) tail, rendering the 3 end open to limited 3 →5
                                                                             exonucleolytic attack [18]. Subsequently, mammalian PABP1
                                                                             was shown to interact in a mutually exclusive manner with
                                                                             eRF3, PAN3 and TOB, and, intriguingly, switching of PABP1
                                                                             from an eRF3-bound to PAN3-bound state requires active
                                                                             translation since it can be blocked by cycloheximide [19].
                                                                             eRF3, PAN3 and TOB all contain a PAM2 motif, and ectopic
                                                                             expression of PAM2-mutant versions of these proteins was
PABP1 function in default mRNA                                               found to exert opposing effects on mRNA deadenylation
deadenylation and decay                                                      rates [19]. Thus, similar to yeast, a model was proposed
                                                                             in which formation of a PABP1–eRF3–eRF1 termination
The dual role of PABP1 in both poly(A) tail                                  complex causes PABP1 to dissociate from the poly(A) tail
protection and deadenylation                                                 (Figure 2, panels 3 and 4). Displaced PABP1 leaves the
The stabilizing effect of a poly(A) tail on a reporter mRNA                  PABP1–eRF3–eRF1 complex upon eRF1 stimulation of
in vitro is negated by sequestration or depletion of PABP1,                  eRF3-mediated GTP hydrolysis [19,20] and it, or the 3 -most
and can be rescued by addition of exogenous PABP1,                           PABP1, may then recruit/stimulate PAN3 or TOB1–CCR4–
with non-adenylated mRNAs being rapidly turned over                          CAF1.
independently of PABP1 status [14]. Since intermediates                          A link between translation termination and incremental
corresponding to the loss of footprints of consecutive PABP                  shortening of the poly(A) tail could provide a molecular
molecules can be observed [15,16], this led to the idea                      timer for mRNA lifespan, limiting the number of times
that PABP1 may act as a steric block to deadenylases                         that an mRNA molecule can be translated [18,19]. However,
(Figure 2, panel 1). Such a function will be determined                      most mRNAs will be translated by many more transiting
by the dissociation rate (K d ) of PABP1 (Figure 2, panel                    ribosomes than would be required to evict the number of
2) which is influenced by whether it is bound non-co-                        PABP1 molecules bound to the poly(A) tail, suggesting that
operatively or co-operatively to poly(A), with the latter                    eviction does not occur at every termination event. Moreover,
exhibiting higher affinity [12]. Mammalian cells contain                     it is unlikely that all default deadenylation is linked to
three major poly(A)-specific 3 exoribonuclease activities:                  termination, since reducing the translational efficiency of
the CCR4–NOT (negative on TATA)–CAF1 complex, the                            a reporter mRNA by introducing a hairpin-loop into the
PAN2–PAN3 complex and PARN [poly(A)-specific                                 5 -UTR (untranslated region) does not block deadenylation
ribonuclease]. Intriguingly, given its poly(A)-protective                    [21]. As discussed above (Figure 2, panel 2), this termination-
effect, PABP1 interacts directly with PARN and the PAN3                      independent deadenylation may simply reflect the off rate
subunit of the PAN2–PAN3 complex and indirectly with                         of PABP1 and/or, since end-to-end complexes may still

                                                                                                
                                                                                                C The   Authors Journal compilation 
                                                                                                                                    C 2012 Biochemical Society
The role of mammalian poly(A)-binding proteins in co-ordinating mRNA turnover
858   Biochemical Society Transactions (2012) Volume 40, part 4

      Figure 2 Role of PABP1 in default deadenylation
                      (1) PABP1 bound to the poly(A) tail prevents access of deadenylases (blunted arrow), but can also recruit and stimulate
                      deadenylases (thin arrow), an activity that is blocked by its interaction with eRF3 which competes for the same binding site
                      within PABC. (2–4) Removal of PABP1 from the poly(A) tail (arrow) facilitates access of deadenylases either as a result of
                      (2) the inherent ‘off’ rate of PABP1 or (3) due to translation termination, since indirect interaction of PABP1 with terminating
                      ribosomes, via an eRF3–eRF1 bridge, causes (4) PABP1 to be evicted from the poly(A) tail. (5) Deadenylated mRNAs
                      are decapped by DCP1–DCP2 (not depicted) and degraded by XRN1 (not depicted), the exosome and/or LSm 1-7–Pat1.
                      TOB1–TOB2 is not depicted.

      form on these mRNAs, PABP1-mediated co-ordination of                             an apparent K d of 150 nM [24], suggesting that, under certain
      deadenylase activity.                                                            circumstances, poly(A) tail-associated PABP1 could directly
                                                                                       protect mRNAs from decapping. However, this binding only
      A direct role for PABP1 in regulating decapping?                                 occurs in the absence of the cap-binding factor eIF4E, thus it
      Deadenylation triggers decapping in mammalian cells ([22]                        is unclear whether this interaction can, or does, occur in cells.
      and reviewed in [23]). Both PABP1 and the poly(A)
      tail are required to protect mRNAs from DCP2-mediated
      decapping in vitro [24,25], suggesting that protection of                        Regulation of aberrant transcript turnover
      the poly(A) tail by PABP1 is likely to be sufficient to                          PTCs (premature termination codons) have been described in
      account for this effect. Intriguingly, however, poly(A)-                         approximately 30 % of known disease-associated mutations
      bound PABP1, but not free PABP1, can bind the RNA-                               [26] and promote aberrant as well as premature translation
      incorporated cap structure, but not free cap, in vitro with                      termination. As a result of their aberrant translation

      
      C The   Authors Journal compilation 
                                          C 2012 Biochemical Society
The role of mammalian poly(A)-binding proteins in co-ordinating mRNA turnover
RNA UK 2012     859

Figure 3 PABP1 suppresses NMD
         (1) mRNAs are initially bound by the nuclear CBC80–CBC20 cap-binding complex with the EJC–UPF2-containing complex
          marking splicing boundaries. As an mRNA emerges from the nucleus, CBC20–CBC80 is exchanged for the cytoplasmic
          cap-binding complex [only eIF4E and eIF4G (4G) are shown], allowing bridging interactions with PABP1. (2) During translation,
          the transiting ribosome displaces the EJC and PABP1–eRF3 complexes promote correct termination ( + ) by interacting with
          eRF1 (not shown). Formation of this complex blocks recruitment of the NMD factor UPF1 (blunted arrow). (3) In contrast,
          when PTCs occur 5 to an EJC, the EJC is not displaced and PABP1 is not proximal to the stop codon (4). Thus normal
          PABP1–eRF3-mediated termination does not occur (blunted arrow) and UPF1 binds the ribosome-associated eRF3, recruiting
          SMG1. (5) The stalled ribosome exits the PTC and the UPF1-containing complex contacts the EJC–UPF2-containing complex
          and inhibits further translation initiation (blunted arrow). (6) The endonuclease SMG6 then cleaves the mRNA 3 to the PTC
          with (7) the mRNA fragments then being degraded in a deadenylation-independent manner. For simplicity, not all NMD
          factors are depicted.

termination, PTC-containing mRNAs can be recognized by                    NMD factors and deadenylation-independent endonuc-
the mRNA-surveillance machinery and subjected to NMD                      leolytic cleavage of the mRNA [27,28]. PABP1 therefore
(nonsense-mediated decay). In mammals, NMD appears to                     plays a role in deciding whether to commit mRNAs for
be triggered by UPF (up-frameshift) 2-containing EJCs                     NMD, since its interaction with eRF3 on the terminating
(exon-junction complexes) located 3 of PTCs. Prematurely                 ribosomes at bona fide stop codons promotes ‘normal’
terminating ribosomes undergo abnormal termination,                       termination and prevents eRF3–UPF1 interaction [29]
with eRF3 being bound by UPF1, which also interacts with                  (Figure 3). The relative proximity of PABP1 to the
EJC-associated UPF2, leading to the recruitment of further                termination codon appears to be important in this decision,

                                                                                                 
                                                                                                 C The   Authors Journal compilation 
                                                                                                                                     C 2012 Biochemical Society
860   Biochemical Society Transactions (2012) Volume 40, part 4

      as NMD can be suppressed by tethering PABP1 in the                 RNA-binding factor 1), NSAP1 (NS1-associated protein 1)
      vicinity of PTCs but 5 to EJCs [30–32]. However, although         and PABP1, bridging a functional interaction between the
      introduction of a PTC which creates a long ‘faux’ (or false)       mCRD and the poly(A) tail and preventing deadenylation
      3 -UTR leads to NMD, many mammalian mRNAs contain                 [40] (Figure 4B). The key interaction in forming this bridging
      long 3 -UTRs and are not subject to NMD [27], underscoring        complex occurs between PABP1 and UNR, although PAIP1
      the importance of a 3 EJC as well as PABP1 proximity.             and AUF1 are also known PABP1-interacting proteins, but
      Nonetheless, increasing the distance between poly(A)-bound         it is unclear whether they interact directly with PABP1
      PABP and a normal stop codon by extending the 3 -UTR              in this context [41]. During c-fos mRNA translation, the
      can induce EJC-independent NMD, which is antagonized by            transiting ribosome displaces the mCRD-bound complex
      reinstating PABP1 proximity [33]. It is not clear how this         [40,41], which may remain associated with poly(A)-bound
      form of NMD is promoted, but one idea is that unknown              PABP1, disrupting the association between the mCRD and
      3 -UTR-associated factors prevent PABP1 from interacting          the poly(A) tail and apparently permitting recruitment of the
      with eRF3 and promote loading of UPF1 to initiate NMD              CCR4 deadenylase by UNR [41]. Thus the translation of c-
      [33].                                                              fos mRNA acts as a trigger for its destruction, thereby limiting
                                                                         the expression of this oncogene by restricting the lifetime of
                                                                         its mRNA.
      mRNA-specific regulation of mRNA
      stability by PABP1                                                 PABP1, AREs and AUBPs
                                                                         Although AUBPs (ARE-binding proteins) can enhance or
      The rate of mRNA decay can be modulated by sequences
                                                                         reduce mRNA stability and/or translation, the presence of
      and/or structural features within mRNAs that, most often,
                                                                         an ARE generally confers reduced basal stability to mRNAs
      recruit specific trans-acting factors (reviewed in [2,23,34]).
                                                                         by enhancing deadenylation [2,42]. Thus it seems likely that
      These cis-acting elements are normally located within
                                                                         there is functional cross-talk between AREs and PABP, and,
      the 3 -UTR as exemplified by AREs (adenine/uridine-rich
                                                                         in support of this, PABP1 associates with AUBPs and can
      elements) and the α-globin mRNA CRE (cytosine-
                                                                         bind to A/U-rich RNA, including functional AREs, in vitro
      rich element) or less frequently, as is the case for c-fos mRNA,
                                                                         [43]. However, despite the large number of identified ARE-
      in the coding region.
                                                                         containing mRNAs and the contribution of misregulated
                                                                         ARE-mediated control to disease (e.g. chronic inflammatory
      PABP1-mediated regulation of endonuclease                          disorders and cancer [34,44,45]), our understanding of the
      recruitment                                                        role of PABPs in ARE-mediated mRNA decay is limited and
      Erythrocytes are enucleated and post-transcriptionally             its role may vary between mRNAs (Figure 4C).
      regulate the synthesis of α- and β-globin subunits for                For instance, an interaction between ARE-bound HuR
      haemoglobin production, with an imbalance in their synthesis       and poly(A)-bound PABP1 was posited to enhance the
      leading to thalassaemias, defective erythropoiesis and/or          ability of PABP1 to protect the poly(A) tail of β-casein
      peripheral haemolysis [35,36]. The 3 -UTR of α-globin             mRNA, contributing to its stability during lactation [46].
      mRNA contains a CRE which is bound by the poly(C)-                 The underlying mechanism remains to be determined, but
      binding protein complex αCP1–αCP2 [15]. Interaction                may reflect altered interaction of PABP1 with poly(A) or
      between PABP1 and αCP1–αCP2 causes an increase in                  deadenylases. In contrast, the AUBP TTP (tristetraprolin)
      both αCP1–αCP2 affinity for the CRE and PABP1-poly(A)              destabilizes mRNA by directly recruiting CAF1 deadenylase
      tail binding, simultaneously blocking ErEN (erythroid-cell-        [47,48], with TTP–PABP1 interactions being reported to
      specific endonuclease) from cleaving at the CRE [15,37] and        inhibit TTP-mediated deadenylation [49]. However, TTP
      reducing deadenylation to stabilize the mRNA (Figure 4A).          dephosphorylation not only enhances its ability to recruit
      A similar mechanism is employed for the stabilization of NF-       CAF1, but also enhances its interaction with PABP1, which
      L (neurofilament light) mRNA, whose misregulation results          appears at odds with its proposed inhibition of TTP-mediated
      in motor neuron degeneration. PABP1 blocks aldolase C-             deadenylation [47]. Thus the functional links between PABP1
      mediated endonucleolytic cleavage within the 3 -UTR of NF-        and TTP await clarification.
      L mRNA, with in vitro studies showing that it binds a 68 nt           PABP1 also interacts with another mRNA-destabilizing
      polypyrimidine-rich region [38,39], implying that PABP1            AUBP, AUF1, whose deficiency in mice leads to the
      may bind to sequences other than poly(A) or A/U.                   stabilization of ARE-containing mRNAs associated with
                                                                         chronic inflammatory diseases [50]. Whereas PABP1 is
      PABP1-mediated regulation of mRNA stability                        proposed to alter TTP function, AUF1 is posited to alter
      via open reading frame cis-acting elements                         PABP1 function. Although the AUF1–PABP1 interaction
      The c-fos mRNA mCRD (major coding-region determinant)              does not directly affect PABP1 poly(A) binding, it was
      is an 87 nt G/A-rich element that directs translationally          proposed that it causes PABP1 eviction from the poly(A)
      coupled deadenylation. The c-fos mCRD binds a mul-                 tail, possibly by modulating the PABP1–eIF4G interaction,
      tiprotein complex containing UNR (upstream of N-ras),              leading to deadenylation. Consistent with this model, Hsp70
      PAIP1 (PABP-interacting protein 1), AUF1 (A/U-rich                 (heat-shock protein 70), which stabilizes ARE-containing

      
      C The   Authors Journal compilation 
                                          C 2012 Biochemical Society
RNA UK 2012     861

Figure 4 Roles of PABP1 in regulating mRNA-specific mRNA turnover
         (A) Left: α-globin mRNA is protected from the action of ErEN and deadenylation (blunted arrows) by 3 -UTR CRE-associated
         αCP1–αCP2 complex which interacts with PABP1, promoting both CRE and poly(A) binding (double-headed arrow). Right:
         in the absence of the αCP1–αCP2 complex binding, ErEN can cleave within the CRE (zigzag arrow), PABP1 poly(A) binding
         will be reduced (broken arrow) and the cleaved mRNA fragments will be subject to decay. (B) Left: on non-translating c-fos
         mRNA, the mCRD is bound by UNR in complex with AUF1, PAIP1 and NSAP1. UNR (and possibly other complex components,
         depicted as broken arrows) interacts with PABP1 and inhibits c-fos mRNA deadenylation (blunted arrow) by an unknown
         mechanism. Right: upon translation of c-fos mRNA, the transiting ribosome displaces the UNR-containing complex from
         the mCRD, although it may remain associated with the poly(A) tail via PABP1 (broken arrow). UNR is then able to recruit
         CCR4–CAF1, and possibly also PAN2–PAN3 (not shown), to promote ( + ) deadenylation. (C) ARE-containing mRNAs are bound
         by one or more AUBPs. AUBPs can promote deadenylation directly through recruitment and stimulation of deadenylases and
         this can be antagonized by PABP1. AUBPs may also promote deadenylation indirectly by altering PABP1 poly(A) binding or
         modulating PABP1-mediated stimulation of deadenylases. Conversely, AUBPs may also inhibit deadenylation by enhancing
         PABP1 poly(A) binding and blocking its ability to stimulate deadenylase activity. Positive and inhibitory actions are depicted by
         + or − respectively. (D) PABP1 function is modulated during miRNA-mediated translational repression and deadenylation.
         RISC (only TNRC6 and Ago shown) is brought to the mRNA via miRNA complementarity. TNRC6 interacts with PABP1 directly
         (shown) and indirectly (not shown) and disrupts its interaction with eIF4G (broken arrow), an interaction which promotes the
         closed-loop conformation and may also enhance PABP1 poly(A) binding (not depicted). This results in repressed translation
         (blunted arrow), facilitating deadenylation. TNRC6 also directly recruits deadenylases and may bring them into close proximity
         to the poly(A) tail via its interaction with PABP1, to promote mRNA deadenylation (arrows). Deadenylase (A and C, see key)
         denotes events in which a specific deadenylase(s) has not been implicated.

                                                                                                  
                                                                                                  C The   Authors Journal compilation 
                                                                                                                                      C 2012 Biochemical Society
862   Biochemical Society Transactions (2012) Volume 40, part 4

      mRNAs [51], disrupts the PABP–AUF1 interaction [51].               PABP in oocytes and early embryos [66,67] and has mainly
      However, ARE binding by AUF1 inhibits its interaction with         has been studied in X. laevis, where it is required for
      PABP1 in vitro [52], suggesting that this interaction does not     both oocyte maturation [68] and early development [64].
      occur on ARE-containing mRNAs that are subject to AUF1-            X. laevis ePABP binds poly(A) [64,66] and has been shown
      mediated deadenylation. Since this observation is inconsistent     to promote mRNA-specific cytoplasmic polyadenylation by
      with an ARE-specific role for this interaction, it raises          protecting newly synthesized poly(A) tails in vitro and in vivo
      questions regarding the role of PABP1 in AUF1-mediated             [68,69] and to prevent deadenylation of ARE-containing
      destabilization. Moreover, AUF1 can also bind poly(A) tails        mRNAs in vitro [66]. It is also likely to inhibit default
      and other components of the closed-loop complex directly           deadenylation via its ability to promote translation initiation
      [52,53], emphasizing further that the role of PABP1 in AUF1-       through eIF4F interaction [67] and translation termin-
      mediated mRNA turnover remains to be fully characterized.          ation through interaction with eRF3 [70]. Recent studies in
                                                                         mice have shown that mammalian ePABP shares the ability
                                                                         to promote cytoplasmic polyadenylation and is required for
      PABP1 and miRNAs (microRNAs)
                                                                         oocyte maturation [71].
      miRNAs play a crucial role in post-transcriptional regulation,
      and their dysregulation is implicated in the aetiology of a
      wide spectrum of human diseases (reviewed in [54]). miRNA-
      mediated silencing utilizes a host of protein factors, including
      the RISC (RNA-induced silencing complex) (reviewed
                                                                         Perspectives
                                                                         Although it is apparent that PABP1 has pleiotropic roles in
      extensively in [55,56]) and PABP1 and is generally considered
                                                                         mRNA turnover, it is also clear that there are currently more
      to involve translational repression and deadenylation. To
                                                                         questions pertaining to those roles than answers. One reason
      date, the order of these events and the extent to which they
                                                                         for the current lack of clarity likely stems from the use of
      promote decay remain controversial.
                                                                         multiple experimental systems, each optimized to analyse
         PABP1 interacts with TNRC6 [56] (Figure 4D), the
                                                                         specific steps in the mRNA translation/turnover pathway.
      RISC component paralogue of GW182, of which there
                                                                         Thus the development of an in vitro system that retains
      are isoforms A–C in mammals. TNRC6 interacts with
                                                                         regulated coupled mRNA translation/turnover would be a
      PABC via a non-canonical PAM2 motif and indirectly
                                                                         boon for studies of post-transcriptional control, and PABPs
      with the RRM region that binds eIF4G, via an unknown
                                                                         in particular. Another reason may be an overreliance on a
      partner [56–58]. The TNRC6–PABP1 interaction is re-
                                                                         small number of reporter mRNAs (e.g. β-globin reporter),
      quired for effective miRNA-mediated silencing [58,59],
                                                                         which are often used in cell types for which they may be
      but, intriguingly, TNRC6 recruits both CCR4–CAF1 and
                                                                         inappropriate.
      PAN2–PAN3 deadenylase complexes directly, and PABP1-
                                                                            A major question pertaining to PABP1 function in
      independently, to effect deadenylation [60–62]. Thus TNRC6
                                                                         default deadenylation relates to understanding the apparent
      may disrupt the PABP1–eIF4G interaction, which is
                                                                         fundamental contradiction between the abilities of poly(A)-
      important for the closed-loop conformation and may also
                                                                         bound PABP1 to both protect an mRNA from deadenylation
      strengthen PABP1 poly(A) binding, to repress translation and
                                                                         and to recruit/enhance deadenylase activities. It is tempting
      enhance deadenylation. Alternatively, TNRC6 may utilize
                                                                         to speculate that this may be controlled, at least in part,
      its interaction with PABP1 to bring the poly(A) tail into
                                                                         by competition between PAN2–PAN3, the TOB1–TOB2–
      proximity of its associated deadenylases [63]. These models
                                                                         CCR4–CAF1 complex and other PABC partners, which
      are not mutually exclusive.
                                                                         have differing affinities (e.g. eRF3
RNA UK 2012     863

   Although even less well understood, mRNA-specific                               12 Lin, J., Fabian, M., Sonenberg, N. and Meller, A. (2012) Nanopore
roles for PABP1 in mRNA turnover are fast emerging and                                detachment kinetics of poly(A) binding proteins from RNA molecules
                                                                                      reveals the critical role of C-terminus interactions. Biophys. J. 102,
supported by multiple findings, although such roles remain                            1427–1434
to be demonstrated in vivo. However, a better understanding                        13 Melo, E.O., Dhalia, R., Martins de Sa, C., Standart, N. and de Melo Neto,
of the role of PABP1 in global mRNA turnover may first be                             O.P. (2003) Identification of a C-terminal poly(A)-binding protein
                                                                                      (PABP)–PABP interaction domain: role in co-operative binding to poly(A)
necessary to inform studies of its mRNA-specific functions.                           and efficient cap distal translational repression. J. Biol. Chem. 278,
Thus whether PABP1 contributes to the regulation of                                   46357–46368
specific mRNAs in multiple ways or whether a unifying                              14 Bernstein, P., Peltz, S.W. and Ross, J. (1989) The poly(A)–poly(A)-binding
                                                                                      protein complex is a major determinant of mRNA stability in vitro. Mol.
mechanism exists remains an important question. A final                               Cell. Biol. 9, 659–670
important, and extremely challenging, question pertains                            15 Wang, Z., Day, N., Trifillis, P. and Kiledjian, M. (1999) An mRNA stability
to the contribution of PABP1-mediated regulation of                                   complex functions with poly(A)-binding protein to stabilize mRNA
                                                                                      in vitro. Mol. Cell. Biol. 19, 4552–4560
both default deadenylation/mRNA stabilization and                                  16 Korner, C.G., Wormington, M., Muckenthaler, M., Schneider, S., Dehlin, E.
mRNA-specific decay to mammalian physiology.                                          and Wahle, E. (1998) The deadenylating nuclease (DAN) is involved in
                                                                                      poly(A) tail removal during the meiotic maturation of Xenopus oocytes.
                                                                                      EMBO J. 17, 5427–5437
                                                                                   17 Ezzeddine, N., Chen, C.Y. and Shyu, A.B. (2012) Evidence providing new
Acknowledgements                                                                      insights into TOB-promoted deadenylation and supporting a link
                                                                                      between TOB’s deadenylation-enhancing and anti-proliferative activities.
                                                                                      Mol. Cell. Biol. 32, 1089–1098
We thank the organizers of RNA UK 2012 for organizing an excellent
                                                                                   18 Hoshino, S., Imai, M., Kobayashi, T., Uchida, N. and Katada, T. (1999) The
meeting and past and present members of the Gray laboratory for                       eukaryotic polypeptide chain releasing factor (eRF3/GSPT) carrying the
helpful discussions of this paper.                                                    translation termination signal to the 3 -poly(A) tail of mRNA: direct
                                                                                      association of erf3/GSPT with polyadenylate-binding protein. J. Biol.
                                                                                      Chem. 274, 16677–16680
                                                                                   19 Funakoshi, Y., Doi, Y., Hosoda, N., Uchida, N., Osawa, M., Shimada, I.,
                                                                                      Tsujimoto, M., Suzuki, T., Katada, T. and Hoshino, S. (2007) Mechanism of
Funding                                                                               mRNA deadenylation: evidence for a molecular interplay between
                                                                                      translation termination factor eRF3 and mRNA deadenylases. Genes Dev.
Work in the Gray laboratory is funded by the Medical Research                         21, 3135–3148
                                                                                   20 Hauryliuk, V., Zavialov, A., Kisselev, L. and Ehrenberg, M. (2006) Class-1
Council (transition funding and a Program Grant) and project grants                   release factor eRF1 promotes GTP binding by class-2 release factor eRF3.
from the Biotechnology and Biological Sciences Research Council                       Biochimie 88, 747–757
and Wellcome Trust.                                                                21 Chen, C.Y., Xu, N. and Shyu, A.B. (1995) mRNA decay mediated by two
                                                                                      distinct AU-rich elements from c-fos and granulocyte-macrophage
                                                                                      colony-stimulating factor transcripts: different deadenylation kinetics and
                                                                                      uncoupling from translation. Mol. Cell. Biol. 15, 5777–5788
                                                                                   22 Yamashita, A., Chang, T.C., Yamashita, Y., Zhu, W., Zhong, Z., Chen, C.Y.
References                                                                            and Shyu, A.B. (2005) Concerted action of poly(A) nucleases and
 1 Kahvejian, A., Svitkin, Y.V., Sukarieh, R., M’Boutchou, M.N. and                   decapping enzyme in mammalian mRNA turnover. Nat. Struct. Mol. Biol.
   Sonenberg, N. (2005) Mammalian poly(A)-binding protein is a                        12, 1054–1063
   eukaryotic translation initiation factor, which acts via multiple               23 Franks, T.M. and Lykke-Andersen, J. (2008) The control of mRNA
   mechanisms. Genes Dev. 19, 104–113                                                 decapping and P-body formation. Mol. Cell 32, 605–615
 2 Chen, C.Y. and Shyu, A.B. (2010) Mechanisms of deadenylation-                   24 Khanna, R. and Kiledjian, M. (2004) Poly(A)-binding-protein-mediated
   dependent decay. Wiley Interdiscip. Rev.: RNA 2, 167–183                           regulation of hDcp2 decapping in vitro. EMBO J. 23, 1968–1976
 3 Tharun, S. (2009) Roles of eukaryotic Lsm proteins in the regulation of         25 Wang, Z., Jiao, X., Carr-Schmid, A. and Kiledjian, M. (2002) The hDcp2
   mRNA function. Int. Rev. Cell Mol. Biol. 272, 149–189                              protein is a mammalian mRNA decapping enzyme. Proc. Natl. Acad. Sci.
                                                                                      U.S.A. 99, 12663–12668
 4 Ling, S.H., Qamra, R. and Song, H. (2011) Structural and functional
                                                                                   26 Bhuvanagiri, M., Schlitter, A.M., Hentze, M.W. and Kulozik, A.E. (2010)
   insights into eukaryotic mRNA decapping. Wiley Interdiscip. Rev.: RNA 2,
                                                                                      NMD: RNA biology meets human genetic medicine. Biochem. J. 430,
   193–208
                                                                                      365–377
 5 Lykke-Andersen, S., Tomecki, R., Jensen, T.H. and Dziembowski, A.
                                                                                   27 Muhlemann, O. and Lykke-Andersen, J. (2011) How and where are
   (2011) The eukaryotic RNA exosome: same scaffold but variable
                                                                                      nonsense mRNAs degraded in mammalian cells? RNA Biol. 7, 28–32
   catalytic subunits. RNA Biol. 8, 61–66
                                                                                   28 Schoenberg, D.R. and Maquat, L.E. (2012) Regulation of cytoplasmic
 6 Brook, M., Smith, J.W.S. and Gray, N.K. (2009) The DAZL and PABP
                                                                                      mRNA decay. Nat. Rev. Genet. 13, 246–259
   families: RNA-binding proteins with interrelated roles in translational         29 Maquat, L.E., Hwang, J., Sato, H. and Tang, Y. (2010) CBP80-promoted
   control in oocytes. Reproduction 137, 595–617                                      mRNP rearrangements during the pioneer round of translation,
 7 Jackson, R.J., Hellen, C.U. and Pestova, T.V. (2010) The mechanism of              nonsense-mediated mRNA decay, and thereafter. Cold Spring Harbor
   eukaryotic translation initiation and principles of its regulation. Nat. Rev.      Symp. Quant. Biol. 75, 127–134
   Mol. Cell Biol. 11, 113–127                                                     30 Silva, A.L., Ribeiro, P., Inacio, A., Liebhaber, S.A. and Romao, L. (2008)
 8 Derry, M.C., Yanagiya, A., Martineau, Y. and Sonenberg, N. (2006)                  Proximity of the poly(A)-binding protein to a premature termination
   Regulation of poly(A)-binding protein through PABP-interacting proteins.           codon inhibits mammalian nonsense-mediated mRNA decay. RNA 14,
   Cold Spring Harbor Symp. Quant. Biol. 71, 537–543                                  563–576
 9 Kozlov, G., Menade, M., Rosenauer, A., Nguyen, L. and Gehring, K.               31 Singh, G., Rebbapragada, I. and Lykke-Andersen, J. (2008) A competition
   (2010) Molecular determinants of PAM2 recognition by the MLLE                      between stimulators and antagonists of Upf complex recruitment
   domain of poly(A)-binding protein. J. Mol. Biol. 397, 397–407                      governs human nonsense-mediated mRNA decay. PLoS Biol. 6, e111
10 Siddiqui, N., Mangus, D.A., Chang, T.C., Palermino, J.M., Shyu, A.B. and        32 Ivanov, P.V., Gehring, N.H., Kunz, J.B., Hentze, M.W. and Kulozik, A.E.
   Gehring, K. (2007) Poly(A) nuclease interacts with the C-terminal                  (2008) Interactions between UPF1, eRFs, PABP and the exon junction
   domain of polyadenylate-binding protein domain from poly(A)-binding                complex suggest an integrated model for mammalian NMD pathways.
   protein. J. Biol. Chem. 282, 25067–25075                                           EMBO J. 27, 736–747
11 Tritschler, F., Huntzinger, E. and Izaurralde, E. (2010) Role of GW182          33 Eberle, A.B., Stalder, L., Mathys, H., Orozco, R.Z. and Muhlemann, O.
   proteins and PABPC1 in the miRNA pathway: a sense of déjà vu. Nat.               (2008) Posttranscriptional gene regulation by spatial rearrangement of
   Rev. Mol. Cell Biol. 11, 379–384                                                   the 3 untranslated region. PLoS Biol. 6, e92

                                                                                                           
                                                                                                           C The   Authors Journal compilation 
                                                                                                                                               C 2012 Biochemical Society
864   Biochemical Society Transactions (2012) Volume 40, part 4

      34 Chatterjee, S. and Pal, J.K. (2009) Role of 5 - and 3 -untranslated regions   54 Esteller, M. (2011) Non-coding RNAs in human disease. Nat. Rev. Genet.
         of mRNAs in human diseases. Biol. Cell 101, 251–262                                12, 861–874
      35 Peixeiro, I., Silva, A.L. and Romao, L. (2011) Control of human β-globin        55 van Kouwenhove, M., Kedde, M. and Agami, R. (2011) MicroRNA
         mRNA stability and its impact on β-thalassemia phenotype.                          regulation by RNA-binding proteins and its implications for cancer. Nat.
         Haematologica 96, 905–913                                                          Rev. 11, 644–656
      36 Voon, H.P. and Vadolas, J. (2008) Controlling α-globin: a review of             56 Huntzinger, E. and Izaurralde, E. (2011) Gene silencing by microRNAs:
         α-globin expression and its impact on β-thalassemia. Haematologica                 contributions of translational repression and mRNA decay. Nat. Rev.
         93, 1868–1876                                                                      Genet. 12, 99–110
      37 Wang, Z. and Kiledjian, M. (2000) The poly(A)-binding protein and an            57 Huntzinger, E., Braun, J.E., Heimstadt, S., Zekri, L. and Izaurralde, E.
         mRNA stability protein jointly regulate an endoribonuclease activity. Mol.         (2010) Two PABPC1-binding sites in GW182 proteins promote
         Cell. Biol. 20, 6334–6341                                                          miRNA-mediated gene silencing. EMBO J. 29, 4146–4160
      38 Stefanizzi, I. and Canete-Soler, R. (2007) Coregulation of light                58 Fabian, M.R., Mathonnet, G., Sundermeier, T., Mathys, H., Zipprich, J.T.,
         neurofilament mRNA by poly(A)-binding protein and aldolase C:                       Svitkin, Y.V., Rivas, F., Jinek, M., Wohlschlegel, J., Doudna, J.A. et al.
         implications for neurodegeneration. Brain Res. 1139, 15–28                         (2009) Mammalian miRNA RISC recruits CAF1 and PABP to affect
      39 Canete-Soler, R., Reddy, K.S., Tolan, D.R. and Zhai, J. (2005) Aldolases A         PABP-dependent deadenylation. Mol. Cell 35, 868–880
         and C are ribonucleolytic components of a neuronal complex that                 59 Jinek, M., Fabian, M.R., Coyle, S.M., Sonenberg, N. and Doudna, J.A.
         regulates the stability of the light-neurofilament mRNA. J. Neurosci. 25,           (2010) Structural insights into the human GW182–PABC interaction in
         4353–4364                                                                          microRNA-mediated deadenylation. Nat. Struct. Mol. Biol. 17, 238–240
      40 Grosset, C., Chen, C.Y., Xu, N., Sonenberg, N., Jacquemin-Sablon, H. and        60 Fabian, M.R., Cieplak, M.K., Frank, F., Morita, M., Green, J., Srikumar, T.,
         Shyu, A.B. (2000) A mechanism for translationally coupled mRNA                     Nagar, B., Yamamoto, T., Raught, B., Duchaine, T.F. et al. (2011)
         turnover: interaction between the poly(A) tail and a c-fos RNA coding              miRNA-mediated deadenylation is orchestrated by GW182 through two
         determinant via a protein complex. Cell 103, 29–40                                 conserved motifs that interact with CCR4-NOT. Nat. Struct. Mol. Biol. 18,
      41 Chang, T.C., Yamashita, A., Chen, C.Y., Yamashita, Y., Zhu, W., Durdan, S.,        1211–1217
         Kahvejian, A., Sonenberg, N. and Shyu, A.B. (2004) UNR, a new partner           61 Braun, J.E., Huntzinger, E., Fauser, M. and Izaurralde, E. (2011) GW182
         of poly(A)-binding protein, plays a key role in translationally coupled            proteins directly recruit cytoplasmic deadenylase complexes to miRNA
         mRNA turnover mediated by the c-fos major coding-region determinant.               targets. Mol. Cell 44, 120–133
         Genes Dev. 18, 2010–2023                                                        62 Chekulaeva, M., Mathys, H., Zipprich, J.T., Attig, J., Colic, M., Parker, R. and
      42 Clark, A., Dean, J., Tudor, C. and Saklatvala, J. (2009) Post-transcriptional      Filipowicz, W. (2011) miRNA repression involves GW182-mediated
         gene regulation by MAP kinases via AU-rich elements. Front. Biosci. 14,            recruitment of CCR4-NOT through conserved W-containing motifs. Nat.
         847–871                                                                            Struct. Mol. Biol. 18, 1218–1226
      43 Sladic, R.T., Lagnado, C.A., Bagley, C.J. and Goodall, G.J. (2004) Human        63 Walters, R.W., Bradrick, S.S. and Gromeier, M. (2010) Poly(A)-binding
         PABP binds AU-rich RNA via RNA-binding domains 3 and 4. Eur. J.                    protein modulates mRNA susceptibility to cap-dependent
         Biochem. 271, 450–457                                                              miRNA-mediated repression. RNA 16, 239–250
      44 Hitti, E. and Khabar, K.S. (2012) Sequence variations affecting AU-rich         64 Gorgoni, B., Richardson, W.A., Burgess, H.M., Anderson, R.C., Wilkie, G.S.,
         element function and disease. Front. Biosci. 17, 1846–1860                         Gautier, P., Martins, J.P., Brook, M., Sheets, M.D. and Gray, N.K. (2011)
      45 Khabar, K.S. (2010) Post-transcriptional control during chronic                    Poly(A)-binding proteins are functionally distinct and have essential roles
         inflammation and cancer: a focus on AU-rich elements. Cell. Mol. Life Sci.          during vertebrate development. Proc. Natl. Acad. Sci. U.S.A. 108,
         67, 2937–2955                                                                      7844–7849
      46 Nagaoka, K., Suzuki, T., Kawano, T., Imakawa, K. and Sakai, S. (2006)           65 Burgess, H.M. and Gray, N.K. (2012) An integrated model for the
         Stability of casein mRNA is ensured by structural interactions between             nucleo-cytoplasmic transport of cytoplasmic poly(A)-binding proteins.
         the 3 -untranslated region and poly(A) tail via the HuR and                       Commun. Integr. Biol. 5, 243–247
         poly(A)-binding protein complex. Biochim. Biophys. Acta 1759,                   66 Voeltz, G.K., Ongkasuwan, J., Standart, N. and Steitz, J.A. (2001) A novel
         132–140                                                                            embryonic poly(A) binding protein, ePAB, regulates mRNA
      47 Marchese, F.P., Aubareda, A., Tudor, C., Saklatvala, J., Clark, A.R. and           deadenylation in Xenopus egg extracts. Genes Dev. 15, 774–788
         Dean, J.L. (2010) MAPKAP kinase 2 blocks tristetraprolin-directed mRNA          67 Wilkie, G.S., Gautier, P., Lawson, D. and Gray, N.K. (2005) Embryonic
         decay by inhibiting CAF1 deadenylase recruitment. J. Biol. Chem. 285,              poly(A)-binding protein stimulates translation in germ cells. Mol. Cell.
         27590–27600                                                                        Biol. 25, 2060–2071
      48 Clement, S.L., Scheckel, C., Stoecklin, G. and Lykke-Andersen, J. (2011)        68 Friend, K., Brook, M., Betül Bezirci, F., Sheets, M., Gray, N.K. and Seli, E.
         Phosphorylation of tristetraprolin by MK2 impairs AU-rich element mRNA             (2012) Embryonic poly(A)-binding protein (ePAB) phosphorylation is
         decay by preventing deadenylase recruitment. Mol. Cell. Biol. 31,                  required for Xenopus oocyte maturation. Biochem. J. 445, 93–100
         256–266                                                                         69 Kim, J.H. and Richter, J.D. (2007) RINGO/cdk1 and CPEB mediate poly(A)
      49 Rowlett, R.M., Chrestensen, C.A., Schroeder, M.J., Harp, M.G., Pelo, J.W.,         tail stabilization and translational regulation by ePAB. Genes Dev. 21,
         Shabanowitz, J., Derose, R., Hunt, D.F., Sturgill, T.W. and Worthington,           2571–2579
         M.T. (2008) Inhibition of tristetraprolin (TTP) deadenylation by poly(A)        70 Cosson, B., Berkova, N., Couturier, A., Chabelskaya, S., Philippe, M. and
         binding protein. Am. J. Physiol. Gastrointest. Liver Physiol. 295,                 Zhouravleva, G. (2002) Poly(A)-binding protein and eRF3 are associated
         G421–G430                                                                          in vivo in human and Xenopus cells. Biol. Cell 94, 205–216
      50 Lu, J.Y., Sadri, N. and Schneider, R.J. (2006) Endotoxic shock in AUF1          71 Guzelogylu-Kayisli, O., Lalioti, M.D., Aydiner, F., Sasson, I., Ilbay, O.,
         knockout mice mediated by failure to degrade proinflammatory cytokine               Sakkas, D., Lowther, K.M., Mehlmann, L.M. and Seli, M. (2012)
         mRNAs. Genes Dev. 20, 3174–3184                                                    Embryonic poly(A)-binding protein (EPAB) is required for oocyte
      51 Laroia, G., Cuesta, R., Brewer, G. and Schneider, R.J. (1999) Control of           maturation and female fertility in mice. Biochem. J. 446, 47–58
         mRNA decay by heat shock–ubiquitin–proteasome pathway. Science                  72 Brook, M., McCracken, L., Reddington, J.P., Lu, Z.L., Morrice, N.A. and
         284, 499–502                                                                       Gray, N.K. (2012) The multifunctional poly(A)-binding protein (PABP) 1 is
      52 Lu, J.Y., Bergman, N., Sadri, N. and Schneider, R.J. (2006) Assembly of            subject to extensive dynamic post-translational modification, which
         AUF1 with eIF4G-poly(A) binding protein complex suggests a translation             molecular modelling suggests plays an important role in co-ordinating its
         function in AU-rich mRNA decay. RNA 12, 883–893                                    activities. Biochem. J. 441, 803–812
      53 Sagliocco, F., Laloo, B., Cosson, B., Laborde, L., Castroviejo, M.,
         Rosenbaum, J., Ripoche, J. and Grosset, C. (2006) The ARE-associated
         factor AUF1 binds poly(A) in vitro in competition with PABP. Biochem. J.        Received 16 April 2012
         400, 337–347                                                                    doi:10.1042/BST20120100

      
      C The   Authors Journal compilation 
                                          C 2012 Biochemical Society
You can also read